1
|
Lu C, Cheng RJ, Zhang Q, Hu Y, Pu Y, Wen J, Zhong Y, Tang Z, Wu L, Wei S, Tsou PS, Fox DA, Li S, Luo Y, Liu Y. Herbal compound cepharanthine attenuates inflammatory arthritis by blocking macrophage M1 polarization. Int Immunopharmacol 2023; 125:111175. [PMID: 37976601 DOI: 10.1016/j.intimp.2023.111175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 10/25/2023] [Accepted: 11/01/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE Cepharanthine (CEP) is a drug candidate for tumor, viral infection, and some inflammatory diseases, but its effect on rheumatoid arthritis (RA) and the underlying mechanism are incompletely understood. METHODS CEP was administered intraperitoneally to a collagen-induced arthritis (CIA) model. Joints went radiological and histological examination and serum cytokines were examined with cytometry-based analysis. M1 macrophages were induced from THP-1 cells or mouse bone marrow-derived macrophages with LPS and IFN-γ. Bulk RNA-seq was performed on macrophage undergoing M1-polarizatioin. Western blotting was applied to determine pathways involved in monocyte chemotaxis and polarization. Glycolysis metabolites were measured by chemiluminescence while glycolytic enzymes were examined by quantitative PCR. RESULTS We found CEP significantly ameliorated synovial inflammation and joint destruction of CIA mice. It downregulated TNF-α levels in serum and in joints. The number of M1 macrophages were reduced in CEP-treated mice. In vitro, CEP inhibited monocyte chemotaxis to MCP-1 by downregulating CCR2 and reducing ERK1/2 signaling. Additionally, CEP suppressed M1 polarization of macrophages induced by LPS and IFN-γ. Genes involved in IFN-γ signaling, IL-6-JAK/STAT3 signaling, glycolysis, and oxidative phosphorylation process were downregulated by CEP. Several enzymes critically involved in glycolytic metabolism were suppressed by CEP, which resulted in reduced citrate in M1-polarizing macrophages. The inhibitory effect of CEP on macrophage polarization might be attributed to the blockage of TLRs-MyD88/IRAK4-IRF5 signaling pathway together with suppression of overactivated glycolytic metabolism in M1-polarizing macrophages. CONCLUSION CEP attenuated joint inflammation by suppressing monocyte chemotaxis and proinflammatory differentiation. It has the potential to be developed into a complementary or alternative therapy for RA.
Collapse
Affiliation(s)
- Chenyang Lu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China; Division of Rheumatology, Department of Internal Medicine, the Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510630, China
| | - Rui-Juan Cheng
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qiuping Zhang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yidan Hu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yaoyu Pu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ji Wen
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yutong Zhong
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhigang Tang
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liang Wu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Shixiong Wei
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, NO. 1 Shuai Fu Yuan, Wang Fu Jing Street, Beijing 100730, China
| | - Pei-Suen Tsou
- Division of Rheumatology, Department of Internal Medicine and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, USA
| | - David A Fox
- Division of Rheumatology, Department of Internal Medicine and Clinical Autoimmunity Center of Excellence, University of Michigan, Ann Arbor, MI, USA
| | - Shasha Li
- Guangdong Provincial Key Laboratory of Diabetology & Guangzhou Municipal Key Laboratory of Mechanistic and Translational Obesity Research, Medical Center for Comprehensive Weight Control, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.
| | - Yubin Luo
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
2
|
Thakur M, Singh M, Kumar S, Dwivedi VP, Dakal TC, Yadav V. A Reappraisal of the Antiviral Properties of and Immune Regulation through Dietary Phytochemicals. ACS Pharmacol Transl Sci 2023; 6:1600-1615. [PMID: 37974620 PMCID: PMC10644413 DOI: 10.1021/acsptsci.3c00178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Indexed: 11/19/2023]
Abstract
In the present era of the COVID-19 pandemic, viral infections remain a major cause of morbidity and mortality worldwide. In this day and age, viral infections are rampant and spreading rapidly. Among the most aggressive viral infections are ebola, AIDS (acquired immunodeficiency syndrome), influenza, and SARS (severe acute respiratory syndrome). Even though there are few treatment options for viral diseases, most of the antiviral therapies are ineffective owing to frequent mutations, the development of more aggressive strains, drug resistance, and possible side effects. Traditionally, herbal remedies have been used by healers, including for dietary and medicinal purposes. Many clinical and scientific studies have demonstrated the therapeutic potential of plant-derived natural compounds. Because of unsafe practices like blood transfusions and organ transplants from infected patients, medical supply contamination. Our antiviral therapies cannot achieve sterile immunity, and we have yet to find a cure for these pernicious infections. Herbs have been shown to improve therapeutic efficacy against a wide variety of viral diseases because of their high concentration of immunomodulatory phytochemicals (both immunoinhibitory and anti-inflammatory). Combined with biotechnology, this folk medicine system can lead to the development of novel antiviral drugs and therapies. In this Review, we will summarize some selected bioactive compounds with probable mechanisms of their antiviral actions, focusing on the immunological axis of these compounds.
Collapse
Affiliation(s)
- Mony Thakur
- Department
of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| | - Mona Singh
- Department
of Obstetrics and Gynaecology, Medical College
of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Sandeep Kumar
- Division
of Cell Biology and Immunology, Council
of Scientific and Industrial Research - Institute of Microbial Technology, Chandigarh 160036, India
| | - Ved Prakash Dwivedi
- International
Centre for Genetic Engineering and Biotechnology, ICGEB Campus, Aruna Asaf Ali Marg, New Delhi 110067, India
| | - Tikam Chand Dakal
- Genome
and Computational Biology Lab, Department of Biotechnology, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001, India
| | - Vinod Yadav
- Department
of Microbiology, Central University of Haryana, Mahendergarh, Haryana 123031, India
| |
Collapse
|
3
|
Jiang G, Xiang Z, Fang Q. Engineering magnetotactic bacteria MVs to synergize chemotherapy, ferroptosis and immunotherapy for augmented antitumor therapy. NANOSCALE HORIZONS 2023; 8:1062-1072. [PMID: 37306000 DOI: 10.1039/d3nh00061c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
One main obstacle to targeted cancer therapies is the immunosuppressive tumor microenvironment, which can facilitate tumor growth and induce resistance to antitumor treatments. Recent studies have indicated that treatment combined with immunotherapy often yields a better prognosis than monotherapy. Bacterial membrane vesicles (MVs), nanostructures released from the membrane of bacteria, can be used as natural nanocarriers for drug delivery and stimulate an immune response because of their immunogenicity. Inspired by the development of synergistic therapeutic strategies, we herein propose a novel nanovaccine-based platform to achieve chemotherapy, ferroptosis therapy, and immunotherapy simultaneously. By simply culturing magnetotactic bacteria in the medium with doxorubicin (DOX) and then extracting specialized MVs (BMVs), BMV@DOX, which are membrane vesicles containing iron ions and DOX, were obtained. We confirmed that in BMV@DOX, the BMV component can stimulate the innate immune system, DOX acts as the chemotherapeutic agent and iron ions will induce ferroptosis. Furthermore, BMV@DOX vesicles modified with DSPE-PEG-cRGD peptides (T-BMV@DOX) have minimized systemic toxicity and increased tumor-specificity. We demonstrated that the smart MVs-based nanovaccine system not only showed superior performance in the treatment of 4T1 breast cancer but also effectively restrained the growth of drug-resistant MCF-7/ADR tumors in mice. Moreover, the nanovaccine could abrogate in vivo lung metastasis of tumor cells in a 4T1-Luc cell induced-lung breast cancer metastasis model. Collectively, the MVs-based nanoplatform offers an alternative promise for surmounting the limitations of monotherapy and may deserve further study for application in synergistic cancer therapy.
Collapse
Affiliation(s)
- Gexuan Jiang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhichu Xiang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Beijing 100190, China.
| | - Qiaojun Fang
- Laboratory of Theoretical and Computational Nanoscience, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Key Laboratory of Nanophotonic Materials and Devices, CAS Center for Excellence in Nanoscience, Beijing Key Laboratory of Ambient Particles Health Effects and Prevention Techniques, National Center for Nanoscience and Technology, Beijing 100190, China.
- University of Chinese Academy of Sciences, Beijing 100049, China
- Sino-Danish Center for Education and Research, Beijing 101408, China
| |
Collapse
|
4
|
Liu K, Hong B, Wang S, Lou F, You Y, Hu R, Shafqat A, Fan H, Tong Y. Pharmacological Activity of Cepharanthine. Molecules 2023; 28:5019. [PMID: 37446681 DOI: 10.3390/molecules28135019] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/16/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Cepharanthine, a natural bisbenzylisoquinoline (BBIQ) alkaloid isolated from the plant Stephania Cephalantha Hayata, is the only bisbenzylisoquinoline alkaloid approved for human use and has been used in the clinic for more than 70 years. Cepharanthine has a variety of medicinal properties, including signaling pathway inhibitory activities, immunomodulatory activities, and antiviral activities. Recently, cepharanthine has been confirmed to greatly inhibit SARS-CoV-2 infection. Therefore, we aimed to describe the pharmacological properties and mechanisms of cepharanthine, mainly including antitumor, anti-inflammatory, anti-pathogen activities, inhibition of bone resorption, treatment of alopecia, treatment of snake bite, and other activities. At the same time, we analyzed and summarized the potential antiviral mechanism of cepharanthine and concluded that one of the most important anti-viral mechanisms of cepharanthine may be the stability of plasma membrane fluidity. Additionally, we explained its safety and bioavailability, which provides evidence for cepharanthine as a potential drug for the treatment of a variety of diseases. Finally, we further discuss the potential new clinical applications of cepharanthine and provide direction for its future development.
Collapse
Affiliation(s)
- Ke Liu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Bixia Hong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Shuqi Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Fuxing Lou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yecheng You
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ruolan Hu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Amna Shafqat
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
5
|
Lu C, Zheng J, Ding Y, Meng Y, Tan F, Gong W, Chu X, Kong X, Gao C. Cepharanthine loaded nanoparticles coated with macrophage membranes for lung inflammation therapy. Drug Deliv 2021; 28:2582-2593. [PMID: 34866533 PMCID: PMC8654408 DOI: 10.1080/10717544.2021.2009936] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Acute lung injury (ALI) is a disease associated with suffering and high lethality, but to date without any effective pharmacological management in the clinic. In the pathological mechanisms of ALI, a strong inflammatory response plays an important role. Herein, based on macrophage 'homing' into inflammation sites and cell membrane coating nanotechnology, we developed a biomimetic anti-inflammation nanosystem (MM-CEP/NLCs) for the treatment of ALI. MM-CEP/NLCs were made with nanostructured lipid carriers (NLCs) coated with natural macrophage membranes (MMs) to achieve effective accumulation of cepharanthine (CEP) in lung inflammation to achieve the effect of treating ALI. With the advantage of suitable physicochemical properties of NLCs and unique biological functions of the macrophage membrane, MM-CEP/NLCs were stabilized and enabled sustained drug release, providing improved biocompatibility and long-term circulation. In vivo, the macrophage membranes enabled NLCs to be targeted and accumulated in the inflammation sites. Further, MM-CEP/NLCs significantly attenuated the severity of ALI, including lung water content, histopathology, bronchioalveolar lavage cellularity, protein concentration, and inflammation cytokines. Our results provide a bionic strategy via the biological properties of macrophages, which may have greater value and application prospects in the treatment of inflammation.
Collapse
Affiliation(s)
- Caihong Lu
- School of Pharmacy, Guangxi Medical University, Nanning, P. R. China.,State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, P. R. China
| | - Jinpeng Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, P. R. China
| | - Yaning Ding
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Shenyang, P. R. China
| | - Yuanyuan Meng
- School of Pharmacy, Guangxi Medical University, Nanning, P. R. China
| | - Fangyun Tan
- School of Pharmacy, Guangxi Medical University, Nanning, P. R. China
| | - Wei Gong
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, P. R. China
| | - Xiaoyang Chu
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing, P. R. China
| | - Xiaolong Kong
- School of Pharmacy, Guangxi Medical University, Nanning, P. R. China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, P. R. China
| |
Collapse
|
6
|
A molecular docking study: Cepharanthine protects articular cartilage against arthritis by Wnt/PI3K/TLR-3 signaling. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2020.10.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
7
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 280] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
8
|
Bailly C. Cepharanthine: An update of its mode of action, pharmacological properties and medical applications. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 62:152956. [PMID: 31132753 PMCID: PMC7126782 DOI: 10.1016/j.phymed.2019.152956] [Citation(s) in RCA: 110] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 05/09/2023]
Abstract
BACKGROUND Cepharanthine (CEP) is a drug used in Japan since the 1950s to treat a number of acute and chronic diseases, including treatment of leukopenia, snake bites, xerostomia and alopecia. It is the only approved drug for Human use in the large class of bisbenzylisoquinoline alkaloids. This natural product, mainly isolated from the plant Stephania cephalantha Hayata, exhibits multiple pharmacological properties including anti-oxidative, anti-inflammatory, immuno-regulatory, anti-cancer, anti-viral and anti-parasitic properties. PURPOSE The mechanism of action of CEP is multifactorial. The drug exerts membrane effects (modulation of efflux pumps, membrane rigidification) as well as different intracellular and nuclear effects. CEP interferes with several metabolic axes, primarily with the AMP-activated protein kinase (AMPK) and NFκB signaling pathways. In particular, the anti-inflammatory effects of CEP rely on AMPK activation and NFκB inhibition. CONCLUSION In this review, the historical discovery and development of CEP are retraced, and the key mediators involved in its mode of action are presented. The past, present, and future of CEP are recapitulated. This review also suggests new opportunities to extend the clinical applications of this well-tolerated old Japanese drug.
Collapse
Affiliation(s)
- Christian Bailly
- UMR-S 1172, Centre de Recherche Jean-Pierre Aubert, INSERM, University of Lille, CHU Lille, 59045, Lille, France; OncoWitan, Lille, Wasquehal, France.
| |
Collapse
|
9
|
Zhou P, Li Z, Xu D, Wang Y, Bai Q, Feng Y, Su G, Chen P, Wang Y, Liu H, Wang X, Zhang R, Wang Y. Cepharanthine Hydrochloride Improves Cisplatin Chemotherapy and Enhances Immunity by Regulating Intestinal Microbes in Mice. Front Cell Infect Microbiol 2019; 9:225. [PMID: 31293986 PMCID: PMC6606789 DOI: 10.3389/fcimb.2019.00225] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 06/11/2019] [Indexed: 12/23/2022] Open
Abstract
Chemotherapy is one of the major treatment strategies for esophageal squamous cell carcinoma (ESCC). Unfortunately, most chemotherapeutic drugs have significant impacts on the intestinal microbes, resulting in side effects and reduced efficiency. Therefore, new strategies capable of overcoming these disadvantages of current chemotherapies are in urgent need. The natural product, Cepharanthine hydrochloride (CEH), is known for its anticancer and immunoregulatory properties. By sequencing the V4 region of 16S rDNA, we characterized the microbes of tumor-bearing mice treated with different chemotherapy strategies, including with CEH. We found that CEH improved the therapeutic effect of CDDP by manipulating the gut microbiota. Through metagenomic analyses of the microbes community, we identified a severe compositional and functional imbalance in the gut microbes community after CDDP treatment. However, CEH improved the effect of chemotherapy and ameliorated CDDP treatment-induced imbalance in the intestinal microbes. Mechanically, CEH activated TLR4 and MYD88 innate immune signaling, which is advantageous for the activation of the host's innate immunity to exert a balanced intestinal environment as well as to trigger a better chemotherapeutic response to esophageal cancer. In addition, TNFR death receptors were activated to induce apoptosis. In summary, our findings suggest that chemotherapy of CDDP combined with CEH increased the effect of chemotherapy and reduced the side effects on the microbes and intestinal mucosal immunity. We believe that these findings provide a theoretical basis for new clinical treatment strategies.
Collapse
Affiliation(s)
- Pengjun Zhou
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Ziyao Li
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Dandan Xu
- Guangdong Food and Drug Vocational College, Guangzhou, China
| | - Ying Wang
- College of Food Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China
| | - Qi Bai
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yue Feng
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Guifeng Su
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Pengxiao Chen
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yao Wang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Huizhong Liu
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xiaogang Wang
- Department of Mathematics and Statistics, York University, Toronto, ON, Canada
| | - Rong Zhang
- State Key Laboratory of Oncology in South China and Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yifei Wang
- Guangdong Provincial Key Laboratory of Bioengineering Medicine, Guangzhou Jinan Biomedicine Research and Development Center, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
10
|
Ren J, Zhao D, Wu SJ, Wang J, Jia YJ, Li WX, Zhu HJ, Cao F, Li W, Pittman CU, He XJ. Reassigning the stereochemistry of bioactive cepharanthine using calculated versus experimental chiroptical spectroscopies. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.01.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
11
|
Uto T, Toyama M, Yoshinaga K, Baba M. Cepharanthine induces apoptosis through the mitochondria/caspase pathway in murine dendritic cells. Immunopharmacol Immunotoxicol 2016; 38:238-43. [PMID: 27121492 DOI: 10.3109/08923973.2016.1173059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
CONTEXT Cepharanthine (CEP) is a biscoclaurine amphipathic alkaloid isolated from the plant Stephania cepharantha Hayata. Although the effects of CEP on several types of cells have been investigated, those on dendritic cells (DCs) are poorly understood. OBJECTIVE To investigate the effect of CEP on the induction of apoptosis in murine DCs. MATERIALS AND METHODS The induction of Annexin V/propidium iodide-positive cells and permeability of mitochondrial membrane potential were evaluated in DCs treated with CEP. Cell-associated caspase activity and DNA fragmentation were analyzed by Dual Sensor: MitoCasp™ and agarose gel electrophoresis, respectively. RESULTS The number of dead cells was increased by CEP treatment at concentrations more than 10 μg/ml. Flow cytometric analysis revealed that the cell death was found to be apoptosis, CEP treatment reduced mitochondrial membrane potential and upregulated the level of cleaved caspases, including caspase-9 and caspase-3/7, in a dose-dependent fashion. Furthermore, DNA fragmentation was observed in CEP-treated DCs. CONCLUSION CEP is capable of inducing apoptosis and may be a potential agent against DC-mediated and allergic diseases.
Collapse
Affiliation(s)
- Tomofumi Uto
- a Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases , Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima , Japan ;,b The Japan Science and Technology Agency (JST), Core Research for Evolutional Science and Technology (CREST) , Tokyo , Japan
| | - Masaaki Toyama
- a Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases , Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima , Japan
| | - Keisuke Yoshinaga
- c Department of Biological and Chemical Systems Engineering , National Institute of Technology, Kumamoto College , Yatsushiro , Japan
| | - Masanori Baba
- a Division of Antiviral Chemotherapy, Center for Chronic Viral Diseases , Graduate School of Medical and Dental Sciences, Kagoshima University , Kagoshima , Japan ;,b The Japan Science and Technology Agency (JST), Core Research for Evolutional Science and Technology (CREST) , Tokyo , Japan
| |
Collapse
|
12
|
Li CW, Menconi F, Osman R, Mezei M, Jacobson EM, Concepcion E, David CS, Kastrinsky DB, Ohlmeyer M, Tomer Y. Identifying a Small Molecule Blocking Antigen Presentation in Autoimmune Thyroiditis. J Biol Chem 2016; 291:4079-90. [PMID: 26703475 PMCID: PMC4759184 DOI: 10.1074/jbc.m115.694687] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/17/2015] [Indexed: 11/06/2022] Open
Abstract
We previously showed that an HLA-DR variant containing arginine at position 74 of the DRβ1 chain (DRβ1-Arg74) is the specific HLA class II variant conferring risk for autoimmune thyroid diseases (AITD). We also identified 5 thyroglobulin (Tg) peptides that bound to DRβ1-Arg74. We hypothesized that blocking the binding of these peptides to DRβ1-Arg74 could block the continuous T-cell activation in thyroiditis needed to maintain the autoimmune response to the thyroid. The aim of the current study was to identify small molecules that can block T-cell activation by Tg peptides presented within DRβ1-Arg74 pockets. We screened a large and diverse library of compounds and identified one compound, cepharanthine that was able to block peptide binding to DRβ1-Arg74. We then showed that Tg.2098 is the dominant peptide when inducing experimental autoimmune thyroiditis (EAT) in NOD mice expressing human DRβ1-Arg74. Furthermore, cepharanthine blocked T-cell activation by thyroglobulin peptides, in particular Tg.2098 in mice that were induced with EAT. For the first time we identified a small molecule that can block Tg peptide binding and presentation to T-cells in autoimmune thyroiditis. If confirmed cepharanthine could potentially have a role in treating human AITD.
Collapse
Affiliation(s)
| | | | - Roman Osman
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Mihaly Mezei
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | | | | | - Chella S David
- the Department of Immunology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, and
| | - David B Kastrinsky
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Michael Ohlmeyer
- Department of Structural and Chemical Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029
| | - Yaron Tomer
- From the Division of Endocrinology, the Bronx Veterans Affairs Medical Center, Bronx, New York 10468
| |
Collapse
|
13
|
Liu Y, Xie D, Kang Y, Wang Y, Yang P, Guo J, Huang J. Microwave-Assisted Extraction Followed by Solid-Phase Extraction for the Chromatographic Analysis of Alkaloids in Stephania cepharantha. J Chromatogr Sci 2016; 54:670-6. [PMID: 26759486 DOI: 10.1093/chromsci/bmv191] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Indexed: 01/07/2023]
Abstract
A procedure involving microwave-assisted extraction (MAE) followed by solid-phase extraction (SPE) was established for the extraction and purification of three bisbenzylisoquinoline alkaloids from Stephania cepharantha, and a reversed-phase high-performance liquid chromatography (HPLC) method was developed for the quantification of the target alkaloids. Chromatographic separation was achieved on a Phenomenex Luna Phenyl-Hexyl column. Prior to the HPLC analysis, the alkaloids were rapidly extracted by an optimized MAE process using 0.01 mol/L hydrochloric acid as the solvent. The MAE extract was subsequently purified by SPE using a cation-exchange polymeric cartridge. The MAE-SPE procedure extracted the three alkaloids with satisfactory recoveries ranging from 100.44 to 102.12%. In comparison with the MAE, Soxhlet and ultrasonic-assisted extractions, the proposed MAE-SPE method showed satisfactory cleanup efficiency. Thus, the validated MAE-SPE-HPLC method is specific, accurate and applicable to the determination of alkaloids in S. cepharantha.
Collapse
Affiliation(s)
- Ying Liu
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Daotao Xie
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Yun Kang
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Yaqin Wang
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Ping Yang
- Instrumental Analysis Center, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Jixian Guo
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| | - Jianming Huang
- Department of Pharmacognosy, School of Pharmacy, Fudan University, Shanghai 201203, PR China
| |
Collapse
|
14
|
Kao MC, Yang CH, Sheu JR, Huang CJ. Cepharanthine mitigates pro-inflammatory cytokine response in lung injury induced by hemorrhagic shock/resuscitation in rats. Cytokine 2015; 76:442-448. [PMID: 26375521 DOI: 10.1016/j.cyto.2015.09.008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 09/02/2015] [Accepted: 09/08/2015] [Indexed: 12/13/2022]
Abstract
BACKGROUND Cepharanthine possesses strong anti-inflammation capacity. We sought to clarify whether cepharanthine could mitigate pro-inflammatory cytokine production in acute lung injury induced by hemorrhagic shock/resuscitation (HS/RES). The involvement of heme oxygenase-1 (HO-1) was also investigated. METHODS Male Sprague Dawley rats were allocated to receive HS/RES, HS/RES plus iv cepharanthine or HS/RES plus cepharanthine plus the HO-1 activity inhibitor tin protoporphyrin (SnPP) and denoted as the HS/RES, HS/RES+CEP, and HS/RES+CEP+SnPP group, respectively. HS/RES was achieved by blood drawing to lower mean arterial pressure (40-45 mmHg for 60 min) followed by shed blood/saline mixtures re-infusion. The rats were monitored for another 5h before sacrifice. RESULTS Arterial blood gas, lung permeability and histologic assays (including histopathology, neutrophil infiltration, and lung water content) confirmed that HS/RES induced significant lung injury. Significant increases in pulmonary levels of tumor necrosis factor-α, interleukin-1β, interleukin-6, prostaglandin E2 and cyclooxygenase-2 confirmed that HS/RES induced a significant inflammatory response in the lungs. Cepharanthine significantly attenuated the pulmonary pro-inflammatory cytokine production and lung injury induced by HS/RES. However, the protective effects of cepharanthine were blocked by SnPP, the potent HO-1 activity inhibitor. CONCLUSION Cepharanthine significantly mitigates pro-inflammatory cytokine response in acute lung injury induced by HS/RES in rats. The mechanism may involve the HO-1 pathway.
Collapse
Affiliation(s)
- Ming-Chang Kao
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Taipei Tzu Chi Hospital, Taipei, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan.
| | - Chen-Hsien Yang
- Department of Anesthesiology, Taipei Tzu Chi Hospital, Taipei, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan.
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Chun-Jen Huang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Anesthesiology, Taipei Tzu Chi Hospital, Taipei, Taiwan; School of Medicine, Tzu Chi University, Hualien, Taiwan.
| |
Collapse
|
15
|
Desgrouas C, Taudon N, Bun SS, Baghdikian B, Bory S, Parzy D, Ollivier E. Ethnobotany, phytochemistry and pharmacology of Stephania rotunda Lour. JOURNAL OF ETHNOPHARMACOLOGY 2014; 154:537-563. [PMID: 24768769 DOI: 10.1016/j.jep.2014.04.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 04/11/2014] [Accepted: 04/12/2014] [Indexed: 06/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Stephania rotunda Lour. (Menispermaceae) is an important traditional medicinal plant that is grown in Southeast Asia. The stems, leaves, and tubers have been used in the Cambodian, Lao, Indian and Vietnamese folk medicine systems for years to treat a wide range of ailments, including asthma, headache, fever, and diarrhoea. AIM OF THE REVIEW To provide an up-to-date, comprehensive overview and analysis of the ethnobotany, phytochemistry, and pharmacology of Stephania rotunda for its potential benefits in human health, as well as to assess the scientific evidence of traditional use and provide a basis for future research directions. MATERIAL AND METHODS Peer-reviewed articles on Stephania rotunda were acquired via an electronic search of the major scientific databases (Pubmed, Google Scholar, and ScienceDirect). Data were collected from scientific journals, theses, and books. RESULTS The traditional uses of Stephania rotunda were recorded in countries throughout Southeast Asia (Cambodia, Vietnam, Laos, and India). Different parts of Stephania rotunda were used in traditional medicine to treat about twenty health disorders. Phytochemical analyses identified forty alkaloids. The roots primarily contain l-tetrahydropalmatine (l-THP), whereas the tubers contain cepharanthine and xylopinine. Furthermore, the chemical composition differs from one region to another and according to the harvest period. The alkaloids exhibited approximately ten different pharmacological activities. The main pharmacological activities of Stephania rotunda alkaloids are antiplasmodial, anticancer, and immunomodulatory effects. Sinomenine, cepharanthine, and l-stepholidine are the most promising components and have been tested in humans. The pharmacokinetic parameters have been studied for seven compounds, including the three most promising compounds. The toxicity has been evaluated for liriodenine, roemerine, cycleanine, l-tetrahydropalmatine, and oxostephanine. CONCLUSION Stephania rotunda is traditionally used for the treatment of a wide range of ailments. Pharmacological investigations have validated different uses of Stephania rotunda in folk medicine. The present review highlights the three most promising compounds of Stephania rotunda, which could constitute potential leads in various medicinal fields, including malaria and cancer.
Collapse
Affiliation(s)
- Camille Desgrouas
- UMR-MD3, IRBA, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France; UMR-MD3, Laboratoire de Pharmacognosie et Ethnopharmacologie, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| | | | - Sok-Siya Bun
- UMR-MD3, Laboratoire de Pharmacognosie et Ethnopharmacologie, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| | - Beatrice Baghdikian
- UMR-MD3, Laboratoire de Pharmacognosie et Ethnopharmacologie, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| | - Sothavireak Bory
- Faculté de Pharmacie, Université des Sciences de la Santé, no. 73, Monivong Blvd, Daun Penh, Phnom Penh, Cambodia.
| | - Daniel Parzy
- UMR-MD3, IRBA, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| | - Evelyne Ollivier
- UMR-MD3, Laboratoire de Pharmacognosie et Ethnopharmacologie, Faculté de Pharmacie, 27 boulevard Jean Moulin CS30064 13385 Marseille cedex 5, Aix-Marseille Université, France.
| |
Collapse
|
16
|
Fu RH, Wang YC, Liu SP, Shih TR, Lin HL, Chen YM, Tsai RT, Tsai CH, Shyu WC, Lin SZ. Dryocrassin Suppresses Immunostimulatory Function of Dendritic Cells and Prolongs Skin Allograft Survival. Cell Transplant 2014; 23:641-56. [PMID: 24816456 DOI: 10.3727/096368914x678373] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Dendritic cells (DCs) are the major specialized antigen-presenting cells for the development of optimal T-cell immunity. DCs can be used as pharmacological targets to monitor novel biological modifiers for the cure of harmful immune responses, such as transplantation rejection. Dryopteris crassirhizoma Nakai (Aspiadaceae) is used for traditional herbal medicine in the region of East Asia. The root of this fern plant has been listed for treating inflammatory diseases. Dryocrassin is the tetrameric phlorophenone component derived from Dryopteris. Here we tested the immunomodulatory potential of dryocrassin on lipopolysaccharide (LPS)-stimulated activation of mouse bone marrow-derived DCs in vitro and in skin allograft transplantation in vivo. Results demonstrated that dryocrassin reduced the emission of tumor necrosis factor-α, interleukin-6, and interleukin-12p70 by LPS-stimulated DCs. The expression of LPS-induced major histocompatibility complex class II, CD40, and CD86 on DCs was also blocked by dryocrassin. Moreover, LPS-stimulated DC-elicited allogeneic T-cell proliferation was alleviated by dryocrassin. In addition, dryocrassin inhibited LPS-induced activation of IkB kinase, JNK/p38 mitogen-activated protein kinase, and the translocation of NF-κB. Treatment with dryocrassin noticeably diminished 2,4-dinitro-1-fluorobenzene-reduced delayed-type hypersensitivity and extended skin allograft survival. Dryocrassin may be one of the potent immunosuppressive agents for transplant rejection via the destruction of DC maturation and function.
Collapse
Affiliation(s)
- Ru-Huei Fu
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | - Yu-Chi Wang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Shih-Ping Liu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Ton-Ru Shih
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Hsin-Lien Lin
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Yue-Mi Chen
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Rong-Tzong Tsai
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Chang-Hai Tsai
- Department of Pediatrics, China Medical University, Taichung, Taiwan
- Department of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Woei-Cherng Shyu
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Department of Neurosurgery, China Medical University Beigang Hospital, Yunlin, Taiwan
- Department of Neurosurgery, Tainan Municipal An-Nan Hospital-China Medical University, Tainan, Taiwan
| |
Collapse
|
17
|
Fu RH, Wang YC, Liu SP, Chu CL, Tsai RT, Ho YC, Chang WL, Chiu SC, Harn HJ, Shyu WC, Lin SZ. Acetylcorynoline impairs the maturation of mouse bone marrow-derived dendritic cells via suppression of IκB kinase and mitogen-activated protein kinase activities. PLoS One 2013; 8:e58398. [PMID: 23472193 PMCID: PMC3589392 DOI: 10.1371/journal.pone.0058398] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2012] [Accepted: 02/04/2013] [Indexed: 12/21/2022] Open
Abstract
Background Dendritic cells (DCs) are major modulators in the immune system. One active field of research is the manipulation of DCs as pharmacological targets to screen novel biological modifiers for the treatment of inflammatory and autoimmune disorders. Acetylcorynoline is the major alkaloid component derived from Corydalis bungeana herbs. We assessed the capability of acetylcorynoline to regulate lipopolysaccharide (LPS)-stimulated activation of mouse bone marrow-derived DCs. Methodology/Principal Findings Our experimental data showed that treatment with up to 20 µM acetylcorynoline does not cause cytotoxicity in cells. Acetylcorynoline significantly inhibited the secretion of tumor necrosis factor-α, interleukin-6, and interleukin-12p70 by LPS-stimulated DCs. The expression of LPS-induced major histocompatibility complex class II, CD40, and CD86 on DCs was also decreased by acetylcorynoline, and the endocytic capacity of LPS-stimulated DCs was restored by acetylcorynoline. In addition, LPS-stimulated DC-elicited allogeneic T-cell proliferation was blocked by acetylcorynoline, and the migratory ability of LPS-stimulated DCs was reduced by acetylcorynoline. Moreover, acetylcorynoline significantly inhibits LPS-induced activation of IκB kinase and mitogen-activated protein kinase. Importantly, administration of acetylcorynoline significantly attenuates 2,4-dinitro-1-fluorobenzene-induced delayed-type hypersensitivity. Conclusions/Significance Acetylcorynoline may be one of the potent immunosuppressive agents through the blockage of DC maturation and function.
Collapse
Affiliation(s)
- Ru-Huei Fu
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- * E-mail: (RHF); (SRL)
| | - Yu-Chi Wang
- Biomedical Technology and Device Research Laboratories, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Shih-Ping Liu
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Ching-Liang Chu
- Graduate Institute of Immunology, National Taiwan University, Taipei, Taiwan
| | - Rong-Tzong Tsai
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yu-Chen Ho
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Wen-Lin Chang
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
| | - Shao-Chih Chiu
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | - Horng-Jyh Harn
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | - Woei-Cherng Shyu
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
| | - Shinn-Zong Lin
- Graduate Institute of Immunology, China Medical University, Taichung, Taiwan
- Center for Neuropsychiatry, China Medical University Hospital, Taichung, Taiwan
- China Medical University Beigang Hospital, Yunlin, Taiwan
- * E-mail: (RHF); (SRL)
| |
Collapse
|